101
|
Sundar R, Brown J, Ingles Russo A, Yap TA. Targeting ATR in cancer medicine. Curr Probl Cancer 2017; 41:302-315. [PMID: 28662958 DOI: 10.1016/j.currproblcancer.2017.05.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/04/2017] [Accepted: 05/15/2017] [Indexed: 12/21/2022]
Abstract
DNA damage occurs continually through various intrinsic and extrinsic mechanisms such as ultraviolet radiation, smoking, reactive oxygen species, and errors during replication. The cellular DNA damage response (DDR) comprises signaling networks that regulate a spectrum of processes, including cell cycle progression, which enable DNA repair to occur. Ataxia telangiectasia mutated (ATM) and ataxia telangiectasia mutated and rad3-related (ATR) kinase are 2 key regulators of the DDR cell cycle checkpoints. ATR plays an essential role in the repair of replication-associated DNA damage, while ATM is activated by DNA double-strand breaks. The investigation of cell cycle checkpoint signaling through ATR and ATM, as well as the relevant pathways involved in oncogenesis and cancer progression, has led to the discovery and development of potent and selective ATR inhibitors (ATRi). Preclinical data have demonstrated that ATR inhibition leads to tumor synthetic lethality in specific molecular contexts, and it exhibits synergy in combination with different antitumor therapies, including chemotherapy, radiotherapy, and poly(ADP-ribose) polymerase inhibitors. ATRi are now being assessed in early-phase clinical trials as single agents and in combinatorial regimens, including platinum and other chemotherapies, radiotherapy, poly(ADP-ribose) polymerase inhibitors, and immune checkpoint inhibitors. This article details the preclinical biology leading to the discovery and development of novel ATRi and discusses the rationale for monotherapy and combination antitumor strategies. We focus on the clinical development of ATRi and discuss the progress made in identifying putative predictive biomarkers of response for patient selection, such as p53, ATM, ARID1A, and other DDR aberrations.
Collapse
Affiliation(s)
- Raghav Sundar
- Drug Development Unit, Royal Marsden Hospital, London, UK; Department of Haematology-Oncology, National University Health System, Singapore
| | - Jessica Brown
- Drug Development Unit, Royal Marsden Hospital, London, UK
| | - Alvaro Ingles Russo
- Drug Development Unit, Royal Marsden Hospital, London, UK; The Institute of Cancer Research, London, UK
| | - Timothy A Yap
- Drug Development Unit, Royal Marsden Hospital, London, UK; The Institute of Cancer Research, London, UK.
| |
Collapse
|
102
|
Targeting the ATR-CHK1 Axis in Cancer Therapy. Cancers (Basel) 2017; 9:cancers9050041. [PMID: 28448462 PMCID: PMC5447951 DOI: 10.3390/cancers9050041] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/23/2017] [Accepted: 04/25/2017] [Indexed: 12/14/2022] Open
Abstract
Targeting the DNA damage response (DDR) is a new therapeutic approach in cancer that shows great promise for tumour selectivity. Key components of the DDR are the ataxia telangiectasia mutated and Rad3 related (ATR) and checkpoint kinase 1 (CHK1) kinases. This review article describes the role of ATR and its major downstream target, CHK1, in the DDR and why cancer cells are particularly reliant on the ATR-CHK1 pathway, providing the rationale for targeting these kinases, and validation of this hypothesis by genetic manipulation. The recent development of specific inhibitors and preclinical data using these inhibitors not only as chemosensitisers and radiosensitisers but also as single agents to exploit specific pathologies of tumour cells is described. These potent and specific inhibitors have now entered clinical trial and early results are presented.
Collapse
|
103
|
Lu Y, Knapp M, Crawford K, Warne R, Elling R, Yan K, Doyle M, Pardee G, Zhang L, Ma S, Mamo M, Ornelas E, Pan Y, Bussiere D, Jansen J, Zaror I, Lai A, Barsanti P, Sim J. Rationally Designed PI3Kα Mutants to Mimic ATR and Their Use to Understand Binding Specificity of ATR Inhibitors. J Mol Biol 2017; 429:1684-1704. [PMID: 28433539 DOI: 10.1016/j.jmb.2017.04.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/08/2017] [Accepted: 04/11/2017] [Indexed: 12/16/2022]
Abstract
ATR, a protein kinase in the PIKK family, plays a critical role in the cell DNA-damage response and is an attractive anticancer drug target. Several potent and selective inhibitors of ATR have been reported showing significant antitumor efficacy, with most advanced ones entering clinical trials. However, due to the absence of an experimental ATR structure, the determinants contributing to ATR inhibitors' potency and specificity are not well understood. Here we present the mutations in the ATP-binding site of PI3Kα to progressively transform the pocket to mimic that of ATR. The generated PI3Kα mutants exhibit significantly improved affinity for selective ATR inhibitors in multiple chemical classes. Furthermore, we obtained the X-ray structures of the PI3Kα mutants in complex with the ATR inhibitors. The crystal structures together with the analysis on the inhibitor affinity profile elucidate the roles of individual amino acid residues in the binding of ATR inhibitors, offering key insights for the binding mechanism and revealing the structure features important for the specificity of ATR inhibitors. The ability to obtain structural and binding data for these PI3Kα mutants, together with their ATR-like inhibitor binding profiles, makes these chimeric PI3Kα proteins valuable model systems for structure-based inhibitor design.
Collapse
Affiliation(s)
- Yipin Lu
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA.
| | - Mark Knapp
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA.
| | - Kenneth Crawford
- Oncology, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Robert Warne
- Oncology, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Robert Elling
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Kelly Yan
- Oncology, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Michael Doyle
- Oncology, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Gwynn Pardee
- Oncology, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Li Zhang
- Oncology, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Sylvia Ma
- Oncology, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Mulugeta Mamo
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Elizabeth Ornelas
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Yue Pan
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Dirksen Bussiere
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Johanna Jansen
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Isabel Zaror
- Oncology, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Albert Lai
- Oncology, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Paul Barsanti
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| | - Janet Sim
- Oncology, Novartis Institutes for Biomedical Research, Emeryville, CA 94608, USA
| |
Collapse
|
104
|
Liu S, Ge Y, Wang T, Edwards H, Ren Q, Jiang Y, Quan C, Wang G. Inhibition of ATR potentiates the cytotoxic effect of gemcitabine on pancreatic cancer cells through enhancement of DNA damage and abrogation of ribonucleotide reductase induction by gemcitabine. Oncol Rep 2017; 37:3377-3386. [PMID: 28440428 DOI: 10.3892/or.2017.5580] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 03/31/2017] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer is a highly malignant disease with a dismal prognosis. Gemcitabine (GEM)-based chemotherapy is the first-line treatment for patients with advanced disease, although its efficacy is very limited, mainly due to drug resistance. Ataxia telangiectasia and Rad3-related (ATR) plays a critical role in the DNA damage response (DDR) which has been implicated in GEM resistance. Thus, targeting ATR represents a promising approach to enhance GEM antitumor activity. In the present study, we tested the antitumor activity of AZ20, a novel ATR-selective inhibitor, alone or combined with GEM in 5 pancreatic cancer cell lines. AZ20 treatment of the pancreatic cancer cell lines resulted in growth inhibition, with IC50 values ranging from 0.84 to 2.4 µM, but limited cell death. As expected, treatment of pancreatic cancer cell lines with AZ20 caused decreased phosphorylation of CHK1 (S-345). However, this was accompanied by DNA damage and S and G2/M cell cycle arrest, independent of TP53 gene mutational status. Importantly, combination of AZ20 with GEM resulted in synergistic inhibition of cell growth and cooperative induction of cell death in the pancreatic cancer cell lines. AZ20 significantly increased GEM-induced DNA damage and almost completely abrogated GEM-induced expression of the M2 subunit of ribonucleotide reductase. These findings suggest that inhibition of ATR is a promising strategy to enhance the antitumor activity of GEM for treating pancreatic cancer.
Collapse
Affiliation(s)
- Shuang Liu
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yubin Ge
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Tingting Wang
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Qihang Ren
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yiqun Jiang
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Chengshi Quan
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guan Wang
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
105
|
Ghelli Luserna di Rora’ A, Iacobucci I, Martinelli G. The cell cycle checkpoint inhibitors in the treatment of leukemias. J Hematol Oncol 2017; 10:77. [PMID: 28356161 PMCID: PMC5371185 DOI: 10.1186/s13045-017-0443-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/15/2017] [Indexed: 01/25/2023] Open
Abstract
The inhibition of the DNA damage response (DDR) pathway in the treatment of cancers has recently reached an exciting stage with several cell cycle checkpoint inhibitors that are now being tested in several clinical trials in cancer patients. Although the great amount of pre-clinical and clinical data are from the solid tumor experience, only few studies have been done on leukemias using specific cell cycle checkpoint inhibitors. This review aims to summarize the most recent data found on the biological mechanisms of the response to DNA damages highlighting the role of the different elements of the DDR pathway in normal and cancer cells and focusing on the main genetic alteration or aberrant gene expression that has been found on acute and chronic leukemias. This review, for the first time, outlines the most important pre-clinical and clinical data available on the efficacy of cell cycle checkpoint inhibitors in single agent and in combination with different agents normally used for the treatment of acute and chronic leukemias.
Collapse
Affiliation(s)
| | - I. Iacobucci
- Department of Hematology and Medical Sciences “L. and A. Seràgnoli”, Bologna University, Bologna, Italy
- Present: Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - G. Martinelli
- Department of Hematology and Medical Sciences “L. and A. Seràgnoli”, Bologna University, Bologna, Italy
| |
Collapse
|
106
|
van Kuijk SJ, Parvathaneni NK, Niemans R, van Gisbergen MW, Carta F, Vullo D, Pastorekova S, Yaromina A, Supuran CT, Dubois LJ, Winum JY, Lambin P. New approach of delivering cytotoxic drugs towards CAIX expressing cells: A concept of dual-target drugs. Eur J Med Chem 2017; 127:691-702. [DOI: 10.1016/j.ejmech.2016.10.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 09/28/2016] [Accepted: 10/16/2016] [Indexed: 12/31/2022]
|
107
|
Carlucci G, Carney B, Sadique A, Vansteene A, Tang J, Reiner T. Evaluation of [ 18F]-ATRi as PET tracer for in vivo imaging of ATR in mouse models of brain cancer. Nucl Med Biol 2017; 48:9-15. [PMID: 28157626 DOI: 10.1016/j.nucmedbio.2017.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/28/2016] [Accepted: 01/05/2017] [Indexed: 10/20/2022]
Abstract
RATIONALE Ataxia telangiectasia and Rad3-related (ATR) threonine serine kinase is one of the key elements in orchestrating the DNA damage response (DDR). As such, inhibition of ATR can amplify the effects of chemo- and radiation-therapy, and several ATR inhibitors (ATRi) have already undergone clinical testing in cancer. For more accurate patient selection, monitoring and staging, real-time in vivo imaging of ATR could be invaluable; the development of appropriate imaging agents has remained a major challenge. METHODS 3-amino-N-(4-[18F]phenyl)-6-(4-(methylsulfonyl)phenyl)pyrazine-2-carboxamide ([18F]-ATRi), a close analogue of Ve-821, (a clinical ATRi candidate), was readily accomplished similarly to already established synthetic procedures. Structurally, 18F was introduced at the 4-position of the aromatic ring of Ve-821 for generating a labeled ATR inhibitor. In vitro experiments were conducted in U251 MG glioblastoma cell lines and ex vivo biodistribution were performed in subcutaneous U251 MG xenograft bearing athymic nude mice following microPET imaging. RESULTS [18F]-ATRi has a similar pharmacokinetic profile to that of Ve-821. Using an U251 MG glioblastoma mouse model, we evaluated the in vivo binding efficiency of [18F]-ATRi. Blood and tumor showed a statistically significant difference between mice injected with only the probe or following blocking experiment with Ve-821 (1.48±0.40%ID/g vs. 0.46±0.12%ID/g in tumor and 1.85±0.47%ID/g vs. 0.84±0.3%ID/g in blood respectively). CONCLUSIONS [18F]-ATRi represents the first 18F positron emission tomography (PET) ATR imaging agent, and is designed on a low nanomolar and clinically relevant ATR inhibitor.
Collapse
Affiliation(s)
- Giuseppe Carlucci
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Brandon Carney
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA; Department of Chemistry, Hunter College and PhD Program in Chemistry, The Graduate Center of the City University of New York, New York, NY 10018, USA
| | - Ahmad Sadique
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Axel Vansteene
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jun Tang
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA; Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
108
|
Williamson CT, Miller R, Pemberton HN, Jones SE, Campbell J, Konde A, Badham N, Rafiq R, Brough R, Gulati A, Ryan CJ, Francis J, Vermulen PB, Reynolds AR, Reaper PM, Pollard JR, Ashworth A, Lord CJ. ATR inhibitors as a synthetic lethal therapy for tumours deficient in ARID1A. Nat Commun 2016; 7:13837. [PMID: 27958275 PMCID: PMC5159945 DOI: 10.1038/ncomms13837] [Citation(s) in RCA: 277] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 11/03/2016] [Indexed: 01/01/2023] Open
Abstract
Identifying genetic biomarkers of synthetic lethal drug sensitivity effects provides one approach to the development of targeted cancer therapies. Mutations in ARID1A represent one of the most common molecular alterations in human cancer, but therapeutic approaches that target these defects are not yet clinically available. We demonstrate that defects in ARID1A sensitize tumour cells to clinical inhibitors of the DNA damage checkpoint kinase, ATR, both in vitro and in vivo. Mechanistically, ARID1A deficiency results in topoisomerase 2A and cell cycle defects, which cause an increased reliance on ATR checkpoint activity. In ARID1A mutant tumour cells, inhibition of ATR triggers premature mitotic entry, genomic instability and apoptosis. The data presented here provide the pre-clinical and mechanistic rationale for assessing ARID1A defects as a biomarker of single-agent ATR inhibitor response and represents a novel synthetic lethal approach to targeting tumour cells.
Collapse
Affiliation(s)
- Chris T. Williamson
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London
SW3 6JB, UK
- The Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London
SW3 6JB, UK
| | - Rowan Miller
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London
SW3 6JB, UK
- The Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London
SW3 6JB, UK
| | - Helen N. Pemberton
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London
SW3 6JB, UK
- The Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London
SW3 6JB, UK
| | - Samuel E. Jones
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London
SW3 6JB, UK
- The Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London
SW3 6JB, UK
| | - James Campbell
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London
SW3 6JB, UK
- The Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London
SW3 6JB, UK
| | - Asha Konde
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London
SW3 6JB, UK
- The Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London
SW3 6JB, UK
| | - Nicholas Badham
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London
SW3 6JB, UK
- The Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London
SW3 6JB, UK
| | - Rumana Rafiq
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London
SW3 6JB, UK
- The Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London
SW3 6JB, UK
| | - Rachel Brough
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London
SW3 6JB, UK
- The Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London
SW3 6JB, UK
| | - Aditi Gulati
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London
SW3 6JB, UK
- The Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London
SW3 6JB, UK
| | - Colm J. Ryan
- Systems Biology Ireland, University College Dublin, Dublin
4, Ireland
| | - Jeff Francis
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London
SW3 6JB, UK
- The Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London
SW3 6JB, UK
| | - Peter B. Vermulen
- The Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London
SW3 6JB, UK
- GZA Hospitals Sint-Augustinus, Wilrijk, Belgium and Center for Oncological Research, University of Antwerp, Oosterveldlaan 24, Wilrijk Antwerp
2610, Belgium
| | - Andrew R. Reynolds
- The Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London
SW3 6JB, UK
| | - Philip M. Reaper
- Vertex Pharmaceuticals (Europe) Limited, Milton Park, Abingdon, Oxfordshire
OX14 4RY, UK
| | - John R. Pollard
- Vertex Pharmaceuticals (Europe) Limited, Milton Park, Abingdon, Oxfordshire
OX14 4RY, UK
| | - Alan Ashworth
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London
SW3 6JB, UK
- The Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London
SW3 6JB, UK
| | - Christopher J. Lord
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London
SW3 6JB, UK
- The Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London
SW3 6JB, UK
| |
Collapse
|
109
|
Apte MS, Cooper JP. Life and cancer without telomerase: ALT and other strategies for making sure ends (don't) meet. Crit Rev Biochem Mol Biol 2016; 52:57-73. [PMID: 27892716 DOI: 10.1080/10409238.2016.1260090] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
While most cancer cells rely on telomerase expression/re-activation for linear chromosome maintenance and sustained proliferation, a significant population of cancers (10-15%) employs telomerase-independent strategies, collectively dubbed Alternative Lengthening of Telomeres (ALT). Most ALT cells relax the usual role of telomeres as inhibitors of local homologous recombination while maintaining the ability of telomeres to prohibit local non-homologous end joining reactions. Here we review current concepts surrounding how ALT telomeres achieve this new balance via alterations in chromatin landscape, DNA damage repair processes and handling of telomeric transcription. We also discuss telomerase independent end maintenance strategies utilized by other organisms, including fruitflies and yeasts, to draw parallels and contrasts and highlight additional modes, beyond ALT, that may be available to telomerase-minus cancers. We conclude by commenting on promises and challenges in the development of effective anti-ALT cancer therapies.
Collapse
Affiliation(s)
- Manasi S Apte
- a Laboratory of Biochemistry and Molecular Biology , Center for Cancer Research, National Cancer Institute, NIH , Bethesda , MD , USA
| | - Julia Promisel Cooper
- a Laboratory of Biochemistry and Molecular Biology , Center for Cancer Research, National Cancer Institute, NIH , Bethesda , MD , USA
| |
Collapse
|
110
|
Cellular responses to replication stress: Implications in cancer biology and therapy. DNA Repair (Amst) 2016; 49:9-20. [PMID: 27908669 DOI: 10.1016/j.dnarep.2016.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 11/15/2016] [Indexed: 12/11/2022]
Abstract
DNA replication is essential for cell proliferation. Any obstacles during replication cause replication stress, which may lead to genomic instability and cancer formation. In this review, we summarize the physiological DNA replication process and the normal cellular response to replication stress. We also outline specialized therapies in clinical trials based on current knowledge and future perspectives in the field.
Collapse
|
111
|
Wagner SA, Oehler H, Voigt A, Dalic D, Freiwald A, Serve H, Beli P. ATR inhibition rewires cellular signaling networks induced by replication stress. Proteomics 2016; 16:402-16. [PMID: 26572502 DOI: 10.1002/pmic.201500172] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 10/15/2015] [Accepted: 11/09/2015] [Indexed: 12/27/2022]
Abstract
The slowing down or stalling of replication forks is commonly known as replication stress and arises from multiple causes such as DNA lesions, nucleotide depletion, RNA-DNA hybrids, and oncogene activation. The ataxia telangiectasia and Rad3-related kinase (ATR) plays an essential role in the cellular response to replication stress and inhibition of ATR has emerged as therapeutic strategy for the treatment of cancers that exhibit high levels of replication stress. However, the cellular signaling induced by replication stress and the substrate spectrum of ATR has not been systematically investigated. In this study, we employed quantitative MS-based proteomics to define the cellular signaling after nucleotide depletion-induced replication stress and replication fork collapse following ATR inhibition. We demonstrate that replication stress results in increased phosphorylation of a subset of proteins, many of which are involved in RNA splicing and transcription and have previously not been associated with the cellular replication stress response. Furthermore, our data reveal the ATR-dependent phosphorylation following replication stress and discover novel putative ATR target sites on MCM6, TOPBP1, RAD51AP1, and PSMD4. We establish that ATR inhibition rewires cellular signaling networks induced by replication stress and leads to the activation of the ATM-driven double-strand break repair signaling.
Collapse
Affiliation(s)
- Sebastian A Wagner
- Department of Medicine, Hematology/Oncology, Goethe University, Frankfurt, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hannah Oehler
- Department of Medicine, Hematology/Oncology, Goethe University, Frankfurt, Germany
| | - Andrea Voigt
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Denis Dalic
- Department of Medicine, Hematology/Oncology, Goethe University, Frankfurt, Germany
| | - Anja Freiwald
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Hubert Serve
- Department of Medicine, Hematology/Oncology, Goethe University, Frankfurt, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Petra Beli
- Institute of Molecular Biology (IMB), Mainz, Germany
| |
Collapse
|
112
|
Chemogenetic profiling identifies RAD17 as synthetically lethal with checkpoint kinase inhibition. Oncotarget 2016; 6:35755-69. [PMID: 26437225 PMCID: PMC4742139 DOI: 10.18632/oncotarget.5928] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 09/14/2015] [Indexed: 01/07/2023] Open
Abstract
Chemical inhibitors of the checkpoint kinases have shown promise in the treatment of cancer, yet their clinical utility may be limited by a lack of molecular biomarkers to identify specific patients most likely to respond to therapy. To this end, we screened 112 known tumor suppressor genes for synthetic lethal interactions with inhibitors of the CHEK1 and CHEK2 checkpoint kinases. We identified eight interactions, including the Replication Factor C (RFC)-related protein RAD17. Clonogenic assays in RAD17 knockdown cell lines identified a substantial shift in sensitivity to checkpoint kinase inhibition (3.5-fold) as compared to RAD17 wild-type. Additional evidence for this interaction was found in a large-scale functional shRNA screen of over 100 genotyped cancer cell lines, in which CHEK1/2 mutant cell lines were unexpectedly sensitive to RAD17 knockdown. This interaction was widely conserved, as we found that RAD17 interacts strongly with checkpoint kinases in the budding yeast Saccharomyces cerevisiae. In the setting of RAD17 knockdown, CHEK1/2 inhibition was found to be synergistic with inhibition of WEE1, another pharmacologically relevant checkpoint kinase. Accumulation of the DNA damage marker γH2AX following chemical inhibition or transient knockdown of CHEK1, CHEK2 or WEE1 was magnified by knockdown of RAD17. Taken together, our data suggest that CHEK1 or WEE1 inhibitors are likely to have greater clinical efficacy in tumors with RAD17 loss-of-function.
Collapse
|
113
|
Ferry-Galow KV, Makhlouf HR, Wilsker DF, Lawrence SM, Pfister TD, Marrero AM, Bigelow KM, Yutzy WH, Ji JJ, Butcher DO, Gouker BA, Kummar S, Chen AP, Kinders RJ, Parchment RE, Doroshow JH. The root causes of pharmacodynamic assay failure. Semin Oncol 2016; 43:484-91. [PMID: 27663480 DOI: 10.1053/j.seminoncol.2016.06.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Robust pharmacodynamic assay results are valuable for informing go/no-go decisions about continued development of new anti-cancer agents and for identifying combinations of targeted agents, but often pharmacodynamic results are too incomplete or variable to fulfill this role. Our experience suggests that variable reagent and specimen quality are two major contributors to this problem. Minimizing all potential sources of variability in procedures for specimen collection, processing, and assay measurements is essential for meaningful comparison of pharmacodynamic biomarkers across sample time points. This is especially true in the evaluation of pre- and post-dose tumor biopsies, which suffer from high levels of tumor insufficiency due to variations in biopsy collection techniques and significant specimen heterogeneity within and across patients. Developing methods to assess heterogeneous biopsies is necessary in order to evaluate a majority of tumor biopsies collected for pharmacodynamic biomarker studies. Improved collection devices and standardization of methods are being sought in order to improve the tumor content and quality of tumor biopsies. In terms of reagent variability, we have found that stringent initial reagent qualification and quality control of R&D-grade reagents is critical to minimize lot-to-lot variability and prevent assay failures, especially for clinical pharmacodynamic questions, which often demand assay performance that meets or exceeds clinical diagnostic assay standards. Rigorous reagent specifications and use of appropriate assay quality control methodologies help to ensure consistency between assay runs, laboratories and trials to provide much needed pharmacodynamic insights into the activity of investigational agents.
Collapse
Affiliation(s)
- Katherine V Ferry-Galow
- Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD.
| | - Hala R Makhlouf
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, Rockville, MD
| | - Deborah F Wilsker
- Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Scott M Lawrence
- Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Thomas D Pfister
- Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD
| | | | - Kristina M Bigelow
- Johns Hopkins School of Medicine, Department of Pharmacology and Molecular Sciences, Baltimore, MD
| | - William H Yutzy
- Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Jiuping J Ji
- Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Donna O Butcher
- Pathology/Histotechnology Laboratory, Animal Sciences Program, Leidos Biomedical Research, Frederick National Laboratories, Frederick, MD
| | - Brad A Gouker
- Pathology/Histotechnology Laboratory, Animal Sciences Program, Leidos Biomedical Research, Frederick National Laboratories, Frederick, MD
| | - Shivaani Kummar
- Stanford University School of Medicine, Department of Oncology, Stanford, CA
| | - Alice P Chen
- NCI/DCTD-Early Clinical Trials Development Program, Bethesda, MD
| | - Robert J Kinders
- Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Ralph E Parchment
- Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| |
Collapse
|
114
|
Cifola I, Lionetti M, Pinatel E, Todoerti K, Mangano E, Pietrelli A, Fabris S, Mosca L, Simeon V, Petrucci MT, Morabito F, Offidani M, Di Raimondo F, Falcone A, Caravita T, Battaglia C, De Bellis G, Palumbo A, Musto P, Neri A. Whole-exome sequencing of primary plasma cell leukemia discloses heterogeneous mutational patterns. Oncotarget 2016; 6:17543-58. [PMID: 26046463 PMCID: PMC4627327 DOI: 10.18632/oncotarget.4028] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 05/11/2015] [Indexed: 02/04/2023] Open
Abstract
Primary plasma cell leukemia (pPCL) is a rare and aggressive form of plasma cell dyscrasia and may represent a valid model for high-risk multiple myeloma (MM). To provide novel information concerning the mutational profile of this disease, we performed the whole-exome sequencing of a prospective series of 12 pPCL cases included in a Phase II multicenter clinical trial and previously characterized at clinical and molecular levels. We identified 1, 928 coding somatic non-silent variants on 1, 643 genes, with a mean of 166 variants per sample, and only few variants and genes recurrent in two or more samples. An excess of C > T transitions and the presence of two main mutational signatures (related to APOBEC over-activity and aging) occurring in different translocation groups were observed. We identified 14 candidate cancer driver genes, mainly involved in cell-matrix adhesion, cell cycle, genome stability, RNA metabolism and protein folding. Furthermore, integration of mutation data with copy number alteration profiles evidenced biallelically disrupted genes with potential tumor suppressor functions. Globally, cadherin/Wnt signaling, extracellular matrix and cell cycle checkpoint resulted the most affected functional pathways. Sequencing results were finally combined with gene expression data to better elucidate the biological relevance of mutated genes. This study represents the first whole-exome sequencing screen of pPCL and evidenced a remarkable genetic heterogeneity of mutational patterns. This may provide a contribution to the comprehension of the pathogenetic mechanisms associated with this aggressive form of PC dyscrasia and potentially with high-risk MM.
Collapse
Affiliation(s)
- Ingrid Cifola
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Marta Lionetti
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,Hematology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Eva Pinatel
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Katia Todoerti
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (PZ), Italy
| | - Eleonora Mangano
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | | | - Sonia Fabris
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,Hematology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Laura Mosca
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,Hematology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Vittorio Simeon
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (PZ), Italy
| | - Maria Teresa Petrucci
- Hematology, Department of Cellular Biotechnologies and Hematology, La Sapienza University, Rome, Italy
| | | | - Massimo Offidani
- Hematologic Clinic, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Francesco Di Raimondo
- Department of Biomedical Sciences, Division of Hematology, Ospedale Ferrarotto, University of Catania, Catania, Italy
| | - Antonietta Falcone
- Hematology Unit, IRCCS "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo, Italy
| | - Tommaso Caravita
- Department of Hematology, Ospedale S. Eugenio, Tor Vergata University, Rome, Italy
| | - Cristina Battaglia
- Institute for Biomedical Technologies, National Research Council, Milan, Italy.,Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Gianluca De Bellis
- Institute for Biomedical Technologies, National Research Council, Milan, Italy
| | - Antonio Palumbo
- Division of Hematology, University of Torino, A.O.U. San Giovanni Battista, Torino, Italy
| | - Pellegrino Musto
- Scientific Direction, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (PZ), Italy
| | - Antonino Neri
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,Hematology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
115
|
Drugging ATR: progress in the development of specific inhibitors for the treatment of cancer. Future Med Chem 2016; 7:873-91. [PMID: 26061106 DOI: 10.4155/fmc.15.33] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In this article, we review the ATR inhibitor field from initial pharmacological tools to first-generation clinical candidates with the potential to bring benefit to cancer patients. ATR is a critical part of the cell DNA-damage response. Over the past decade or more, compounds with weak ATR potency and low specificity have been used as tools in early studies to elucidate ATR pharmacology. More recently highly potent, selective and in vivo active ATR inhibitors have been developed enabling detailed preclinical in vitro and in vivo target assessment to be made. The published studies reveal the potential of ATR inhibitors for use as monotherapy or in combination with DNA-damaging agents. To date, VX-970 and AZD6738, have entered clinical assessment.
Collapse
|
116
|
Rocca CJ, Soares DG, Bouzid H, Henriques JAP, Larsen AK, Escargueil AE. BRCA2 is needed for both repair and cell cycle arrest in mammalian cells exposed to S23906, an anticancer monofunctional DNA binder. Cell Cycle 2016; 14:2080-90. [PMID: 25945522 DOI: 10.1080/15384101.2015.1042632] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Repair of DNA-targeted anticancer agents is an active area of investigation of both fundamental and clinical interest. However, most studies have focused on a small number of compounds limiting our understanding of both DNA repair and the DNA damage response. S23906 is an acronycine derivative that shows strong activity toward solid tumors in experimental models. S23906 forms bulky monofunctional DNA adducts in the minor groove which leads to destabilization of the double-stranded helix. We now report that S23906 induces formation of DNA double strand breaks that are processed through homologous recombination (HR) but not Non-Homologous End-Joining (NHEJ) repair. Interestingly, S23906 exposure was accompanied by a higher sensitivity of BRCA2-deficient cells compared to other HR deficient cell lines and by an S-phase accumulation in wild-type (wt), but not in BRCA2-deficient cells. Recently, we have shown that S23906-induced S phase arrest was mediated by the checkpoint kinase Chk1. However, its activated phosphorylated form is equally induced by S23906 in wt and BRCA2-deficient cells, likely indicating a role for BRCA2 downstream of Chk1. Accordingly, override of the S phase arrest by either 7-hydroxystaurosporine (UCN-01) or AZD7762 potentiates the cytotoxic activity of S23906 in wt, but not in BRCA2-deficient cells. Together, our findings suggest that the pronounced sensitivity of BRCA2-deficient cells to S23906 is due to both a defective S-phase arrest and the absence of HR repair. Tumors with deficiencies for proteins involved in HR, and BRCA2 in particular, may thus show increased sensitivity to S23906, thereby providing a rationale for patient selection in clinical trials.
Collapse
Key Words
- ATR, Ataxia telangiectasia- and RAD3-related
- DNA alkylators
- DNA double strand breaks
- DNA replication
- DSBs, Double Strand Breaks
- FA, Fanconi Anemia
- GAPDH, Glyceraldehyde-3-phosphate dehydrogenase
- HR, Homologous Recombination
- HU, Hydroxyurea
- Homologous recombination
- ICLs, Inter-strand Crosslinks
- NER, Nucleotide Excision Repair
- NHEJ, Non-Homologous End-Joining
- TCR, Transcription-Coupled Repair
- UCN-01, 7-hydroxystaurosporine.
- checkpoint control
Collapse
Affiliation(s)
- Céline J Rocca
- a Laboratory of Cancer Biology and Therapeutics ; Centre de Recherche Saint-Antoine ; Paris , France
| | | | | | | | | | | |
Collapse
|
117
|
Abstract
An underlying hallmark of cancers is their genomic instability, which is associated with a greater propensity to accumulate DNA damage. Historical treatment of cancer by radiotherapy and DNA-damaging chemotherapy is based on this principle, yet it is accompanied by significant collateral damage to normal tissue and unwanted side effects. Targeted therapy based on inhibiting the DNA damage response (DDR) in cancers offers the potential for a greater therapeutic window by tailoring treatment to patients with tumors lacking specific DDR functions. The recent approval of olaparib (Lynparza), the poly (ADP-ribose) polymerase (PARP) inhibitor for treating tumors harboring BRCA1 or BRCA2 mutations, represents the first medicine based on this principle, exploiting an underlying cause of tumor formation that also represents an Achilles' heel. This review highlights the different concepts behind targeting DDR in cancer and how this can provide significant opportunities for DDR-based therapies in the future.
Collapse
|
118
|
Kemp MG, Sancar A. ATR Kinase Inhibition Protects Non-cycling Cells from the Lethal Effects of DNA Damage and Transcription Stress. J Biol Chem 2016; 291:9330-42. [PMID: 26940878 DOI: 10.1074/jbc.m116.719740] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Indexed: 01/09/2023] Open
Abstract
ATR (ataxia telangiectasia and Rad-3-related) is a protein kinase that maintains genome stability and halts cell cycle phase transitions in response to DNA lesions that block DNA polymerase movement. These DNA replication-associated features of ATR function have led to the emergence of ATR kinase inhibitors as potential adjuvants for DNA-damaging cancer chemotherapeutics. However, whether ATR affects the genotoxic stress response in non-replicating, non-cycling cells is currently unknown. We therefore used chemical inhibition of ATR kinase activity to examine the role of ATR in quiescent human cells. Although ATR inhibition had no obvious effects on the viability of non-cycling cells, inhibition of ATR partially protected non-replicating cells from the lethal effects of UV and UV mimetics. Analyses of various DNA damage response signaling pathways demonstrated that ATR inhibition reduced the activation of apoptotic signaling by these agents in non-cycling cells. The pro-apoptosis/cell death function of ATR is likely due to transcription stress because the lethal effects of compounds that block RNA polymerase movement were reduced in the presence of an ATR inhibitor. These results therefore suggest that whereas DNA polymerase stalling at DNA lesions activates ATR to protect cell viability and prevent apoptosis, the stalling of RNA polymerases instead activates ATR to induce an apoptotic form of cell death in non-cycling cells. These results have important implications regarding the use of ATR inhibitors in cancer chemotherapy regimens.
Collapse
Affiliation(s)
- Michael G Kemp
- From the Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599
| | - Aziz Sancar
- From the Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599
| |
Collapse
|
119
|
Abstract
Over the past decade, rapid advances in genomics, proteomics and functional genomics technologies that enable in-depth interrogation of cancer genomes and proteomes and high-throughput analysis of gene function have enabled characterization of the kinome 'at large' in human cancers, providing crucial insights into how members of the protein kinase superfamily are dysregulated in malignancy, the context-dependent functional role of specific kinases in cancer and how kinome remodelling modulates sensitivity to anticancer drugs. The power of these complementary approaches, and the insights gained from them, form the basis of this Analysis article.
Collapse
Affiliation(s)
- Emmy D G Fleuren
- Department of Medical Oncology, Radboud University Medical Centre, Geert Grooteplein-Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Luxi Zhang
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
- University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Jianmin Wu
- Cancer Division, Kinghorn Cancer Centre, Garvan Institute of Medical Research, 370 Victoria Street, Sydney, New South Wales 2010, Australia
| | - Roger J Daly
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
120
|
Muralidharan SV, Bhadury J, Nilsson LM, Green LC, McLure KG, Nilsson JA. BET bromodomain inhibitors synergize with ATR inhibitors to induce DNA damage, apoptosis, senescence-associated secretory pathway and ER stress in Myc-induced lymphoma cells. Oncogene 2016; 35:4689-97. [PMID: 26804177 DOI: 10.1038/onc.2015.521] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 11/08/2015] [Accepted: 12/11/2015] [Indexed: 12/31/2022]
Abstract
Inhibiting the bromodomain and extra-terminal (BET) domain family of epigenetic reader proteins has been shown to have potent anti-tumoral activity, which is commonly attributed to suppression of transcription. In this study, we show that two structurally distinct BET inhibitors (BETi) interfere with replication and cell cycle progression of murine Myc-induced lymphoma cells at sub-lethal concentrations when the transcriptome remains largely unaltered. This inhibition of replication coincides with a DNA-damage response and enhanced sensitivity to inhibitors of the upstream replication stress sensor ATR in vitro and in mouse models of B-cell lymphoma. Mechanistically, ATR and BETi combination therapy cause robust transcriptional changes of genes involved in cell death, senescence-associated secretory pathway, NFkB signaling and ER stress. Our data reveal that BETi can potentiate the cell stress and death caused by ATR inhibitors. This suggests that ATRi can be used in combination therapies of lymphomas without the use of genotoxic drugs.
Collapse
Affiliation(s)
- S V Muralidharan
- Department of Surgery, Sahlgrenska Cancer Center, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - J Bhadury
- Department of Surgery, Sahlgrenska Cancer Center, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - L M Nilsson
- Department of Surgery, Sahlgrenska Cancer Center, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - L C Green
- Department of Surgery, Sahlgrenska Cancer Center, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - K G McLure
- Zenith Epigenetics Corp, Calgary, Alberta, Canada
| | - J A Nilsson
- Department of Surgery, Sahlgrenska Cancer Center, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
121
|
Sanjiv K, Hagenkort A, Calderón-Montaño JM, Koolmeister T, Reaper PM, Mortusewicz O, Jacques SA, Kuiper RV, Schultz N, Scobie M, Charlton PA, Pollard JR, Berglund UW, Altun M, Helleday T. Cancer-Specific Synthetic Lethality between ATR and CHK1 Kinase Activities. Cell Rep 2015; 14:298-309. [PMID: 26748709 PMCID: PMC4713868 DOI: 10.1016/j.celrep.2015.12.032] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 11/04/2015] [Accepted: 12/03/2015] [Indexed: 02/05/2023] Open
Abstract
ATR and CHK1 maintain cancer cell survival under replication stress and inhibitors of both kinases are currently undergoing clinical trials. As ATR activity is increased after CHK1 inhibition, we hypothesized that this may indicate an increased reliance on ATR for survival. Indeed, we observe that replication stress induced by the CHK1 inhibitor AZD7762 results in replication catastrophe and apoptosis, when combined with the ATR inhibitor VE-821 specifically in cancer cells. Combined treatment with ATR and CHK1 inhibitors leads to replication fork arrest, ssDNA accumulation, replication collapse, and synergistic cell death in cancer cells in vitro and in vivo. Inhibition of CDK reversed replication stress and synthetic lethality, demonstrating that regulation of origin firing by ATR and CHK1 explains the synthetic lethality. In conclusion, this study exemplifies cancer-specific synthetic lethality between two proteins in the same pathway and raises the prospect of combining ATR and CHK1 inhibitors as promising cancer therapy.
Collapse
Affiliation(s)
- Kumar Sanjiv
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Anna Hagenkort
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - José Manuel Calderón-Montaño
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Tobias Koolmeister
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Philip M Reaper
- Vertex Pharmaceuticals (Europe) Ltd., Abingdon, Oxfordshire OX14 4RW, UK
| | - Oliver Mortusewicz
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Sylvain A Jacques
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Raoul V Kuiper
- Laboratory Medicine, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Niklas Schultz
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Martin Scobie
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Peter A Charlton
- Vertex Pharmaceuticals (Europe) Ltd., Abingdon, Oxfordshire OX14 4RW, UK
| | - John R Pollard
- Vertex Pharmaceuticals (Europe) Ltd., Abingdon, Oxfordshire OX14 4RW, UK
| | - Ulrika Warpman Berglund
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Mikael Altun
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden
| | - Thomas Helleday
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 21 Stockholm, Sweden.
| |
Collapse
|
122
|
Puigvert JC, Sanjiv K, Helleday T. Targeting DNA repair, DNA metabolism and replication stress as anti-cancer strategies. FEBS J 2015; 283:232-45. [DOI: 10.1111/febs.13574] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 10/04/2015] [Accepted: 10/21/2015] [Indexed: 12/16/2022]
Affiliation(s)
- Jordi Carreras Puigvert
- Science for Life Laboratory; Division of Translational Medicine and Chemical Biology; Department of Medical Biochemistry and Biophysics; Karolinska Institutet; Stockholm Sweden
| | - Kumar Sanjiv
- Science for Life Laboratory; Division of Translational Medicine and Chemical Biology; Department of Medical Biochemistry and Biophysics; Karolinska Institutet; Stockholm Sweden
| | - Thomas Helleday
- Science for Life Laboratory; Division of Translational Medicine and Chemical Biology; Department of Medical Biochemistry and Biophysics; Karolinska Institutet; Stockholm Sweden
| |
Collapse
|
123
|
Alsubhi N, Middleton F, Abdel-Fatah TMA, Stephens P, Doherty R, Arora A, Moseley PM, Chan SYT, Aleskandarany MA, Green AR, Rakha EA, Ellis IO, Martin SG, Curtin NJ, Madhusudan S. Chk1 phosphorylated at serine345 is a predictor of early local recurrence and radio-resistance in breast cancer. Mol Oncol 2015; 10:213-23. [PMID: 26459098 DOI: 10.1016/j.molonc.2015.09.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 09/15/2015] [Accepted: 09/19/2015] [Indexed: 12/28/2022] Open
Abstract
Radiation-induced DNA damage activates the DNA damage response (DDR). DDR up-regulation may predict radio-resistance and increase the risk of early local recurrence despite radiotherapy in early stage breast cancers. In 1755 early stage breast cancers, DDR signalling [ATM, ATR, total Ckh1, Chk1 phosphorylated at serine(345) (pChk1), Chk2, p53], base excision repair [PARP1, POLβ, XRCC1, FEN1, SMUG1], non-homologous end joining (Ku70/Ku80, DNA-PKcs) and homologous recombination [RAD51, BRCA1, γH2AX, BLM, WRN, RECQL5, PTEN] protein expression was correlated to time to early local recurrence. Pre-clinically, radio-sensitization by inhibition of Chk1 activation by ATR inhibitor (VE-821) and inhibition of Chk1 (V158411) were investigated in MDA-MB-231 (p53 mutant) and MCF-7 (p53 wild-type) breast cancer cells. In the whole cohort, 208/1755 patients (11.9%) developed local recurrence of which 126 (61%) developed local recurrence within 5 years of initiation of primary therapy. Of the 20 markers tested, only pChk1 and p53 significantly associated with early local recurrence (p value = 0.015 and 0.010, respectively). When analysed together, high cytoplasmic pChk1-nuclear pChk1 (p = 0.039), high cytoplasmic pChk1-p53 (p = 0.004) and high nuclear pChk1-p53 (p = 0.029) co-expression remain significantly linked to early local recurrence. In multivariate analysis, cytoplasmic pChk1 level independently predicted early local recurrence (p = 0.025). In patients who received adjuvant local radiotherapy (n = 949), p53 (p = 0.014) and high cytoplasmic pChk1-p53 (p = 0.017) remain associated with early local recurrence. Pre-clinically, radio-sensitisation by VE-821 or V158411 was observed in both MCF-7 and MDA-MB-231 cells and was more pronounced in MCF-7 cells. We conclude that pChk1 is a predictive biomarker of radiotherapy resistance and early local recurrence.
Collapse
Affiliation(s)
- Nouf Alsubhi
- Academic Unit of Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG51PB, UK
| | - Fiona Middleton
- Northern Institute for Cancer Research, School of Clinical & Laboratory Sciences, Newcastle University, Medical School, Newcastle upon Tyne NE2 4HH, UK
| | | | - Peter Stephens
- Northern Institute for Cancer Research, School of Clinical & Laboratory Sciences, Newcastle University, Medical School, Newcastle upon Tyne NE2 4HH, UK
| | - Rachel Doherty
- Academic Unit of Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG51PB, UK
| | - Arvind Arora
- Academic Unit of Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG51PB, UK
| | - Paul M Moseley
- Department of Oncology, Nottingham University Hospitals, Nottingham NG51PB, UK
| | - Stephen Y T Chan
- Department of Oncology, Nottingham University Hospitals, Nottingham NG51PB, UK
| | | | - Andrew R Green
- Department of Pathology, School of Medicine, University of Nottingham, Nottingham NG51PB, UK
| | - Emad A Rakha
- Department of Pathology, School of Medicine, University of Nottingham, Nottingham NG51PB, UK
| | - Ian O Ellis
- Department of Pathology, School of Medicine, University of Nottingham, Nottingham NG51PB, UK
| | - Stewart G Martin
- Academic Unit of Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG51PB, UK
| | - Nicola J Curtin
- Northern Institute for Cancer Research, School of Clinical & Laboratory Sciences, Newcastle University, Medical School, Newcastle upon Tyne NE2 4HH, UK.
| | - Srinivasan Madhusudan
- Academic Unit of Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG51PB, UK; Department of Oncology, Nottingham University Hospitals, Nottingham NG51PB, UK.
| |
Collapse
|
124
|
Manic G, Obrist F, Sistigu A, Vitale I. Trial Watch: Targeting ATM-CHK2 and ATR-CHK1 pathways for anticancer therapy. Mol Cell Oncol 2015; 2:e1012976. [PMID: 27308506 PMCID: PMC4905354 DOI: 10.1080/23723556.2015.1012976] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 01/25/2015] [Accepted: 01/26/2015] [Indexed: 02/08/2023]
Abstract
The ataxia telangiectasia mutated serine/threonine kinase (ATM)/checkpoint kinase 2 (CHEK2, best known as CHK2) and the ATM and Rad3-related serine/threonine kinase (ATR)/CHEK1 (best known as CHK1) cascades are the 2 major signaling pathways driving the DNA damage response (DDR), a network of processes crucial for the preservation of genomic stability that act as a barrier against tumorigenesis and tumor progression. Mutations and/or deletions of ATM and/or CHK2 are frequently found in tumors and predispose to cancer development. In contrast, the ATR-CHK1 pathway is often upregulated in neoplasms and is believed to promote tumor growth, although some evidence indicates that ATR and CHK1 may also behave as haploinsufficient oncosuppressors, at least in a specific genetic background. Inactivation of the ATM-CHK2 and ATR-CHK1 pathways efficiently sensitizes malignant cells to radiotherapy and chemotherapy. Moreover, ATR and CHK1 inhibitors selectively kill tumor cells that present high levels of replication stress, have a deficiency in p53 (or other DDR players), or upregulate the ATR-CHK1 module. Despite promising preclinical results, the clinical activity of ATM, ATR, CHK1, and CHK2 inhibitors, alone or in combination with other therapeutics, has not yet been fully demonstrated. In this Trial Watch, we give an overview of the roles of the ATM-CHK2 and ATR-CHK1 pathways in cancer initiation and progression, and summarize the results of clinical studies aimed at assessing the safety and therapeutic profile of regimens based on inhibitors of ATR and CHK1, the only 2 classes of compounds that have so far entered clinics.
Collapse
Affiliation(s)
| | - Florine Obrist
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
- INSERM, UMRS1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | | | - Ilio Vitale
- Regina Elena National Cancer Institute; Rome, Italy
- Department of Biology, University of Rome “TorVergata”; Rome, Italy
| |
Collapse
|
125
|
Buisson R, Boisvert JL, Benes CH, Zou L. Distinct but Concerted Roles of ATR, DNA-PK, and Chk1 in Countering Replication Stress during S Phase. Mol Cell 2015; 59:1011-24. [PMID: 26365377 PMCID: PMC4575890 DOI: 10.1016/j.molcel.2015.07.029] [Citation(s) in RCA: 243] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 05/08/2015] [Accepted: 07/28/2015] [Indexed: 01/29/2023]
Abstract
The ATR-Chk1 pathway is critical for DNA damage responses and cell-cycle progression. Chk1 inhibition is more deleterious to cycling cells than ATR inhibition, raising questions about ATR and Chk1 functions in the absence of extrinsic replication stress. Here we show that a key role of ATR in S phase is to coordinate RRM2 accumulation and origin firing. ATR inhibitor (ATRi) induces massive ssDNA accumulation and replication catastrophe in a fraction of early S-phase cells. In other S-phase cells, however, ATRi induces moderate ssDNA and triggers a DNA-PK and Chk1-mediated backup pathway to suppress origin firing. The backup pathway creates a threshold such that ATRi selectively kills cells under high replication stress, whereas Chk1 inhibitor induces cell death at a lower threshold. The levels of ATRi-induced ssDNA correlate with ATRi sensitivity in a panel of cell lines, suggesting that ATRi-induced ssDNA could be predictive of ATRi sensitivity in cancer cells.
Collapse
Affiliation(s)
- Rémi Buisson
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02129, USA
| | - Jessica L Boisvert
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02129, USA
| | - Cyril H Benes
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02129, USA
| | - Lee Zou
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02129, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
126
|
Karnitz LM, Zou L. Molecular Pathways: Targeting ATR in Cancer Therapy. Clin Cancer Res 2015; 21:4780-5. [PMID: 26362996 DOI: 10.1158/1078-0432.ccr-15-0479] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/12/2015] [Indexed: 12/19/2022]
Abstract
The human ATR gene encodes a kinase that is activated by DNA damage and replication stress as a central transducer of a checkpoint signaling pathway. Once activated, ATR phosphorylates multiple substrates, including the kinase Chk1, to regulate cell-cycle progression, replication fork stability, and DNA repair. These events promote cell survival during replication stress and in cells with DNA damage. Accordingly, there has been the tantalizing possibility that ATR inhibitors would be therapeutically useful, especially if they were more effective in tumor versus normal cells. Indeed, multiple studies have demonstrated that alterations that promote tumorigenesis, such as defects in the ATM-p53 pathway, constitutive oncogene activation, and acquisition of the alternative lengthening of telomeres pathway, render tumor cells sensitive to ATR inhibitor monotherapy and/or increase the synergy between ATR inhibitors and genotoxic chemotherapies. Now, nearly two decades after the discovery of ATR, two highly selective and potent ATR inhibitors, AZD6738 and VX-970, are in early-phase clinical trials either as monotherapies or paired with a variety of genotoxic chemotherapies. These trials will generate important insights into the effects of ATR inhibition in humans and the potential role of inhibiting this kinase in the treatment of human malignancies.
Collapse
Affiliation(s)
- Larry M Karnitz
- Division of Oncology Research and Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota.
| | - Lee Zou
- Massachusetts General Hospital Cancer Center, Department of Pathology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
127
|
Xu Y, Her C. Inhibition of Topoisomerase (DNA) I (TOP1): DNA Damage Repair and Anticancer Therapy. Biomolecules 2015; 5:1652-70. [PMID: 26287259 PMCID: PMC4598769 DOI: 10.3390/biom5031652] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 07/14/2015] [Indexed: 12/31/2022] Open
Abstract
Most chemotherapy regimens contain at least one DNA-damaging agent that preferentially affects the growth of cancer cells. This strategy takes advantage of the differences in cell proliferation between normal and cancer cells. Chemotherapeutic drugs are usually designed to target rapid-dividing cells because sustained proliferation is a common feature of cancer [1,2]. Rapid DNA replication is essential for highly proliferative cells, thus blocking of DNA replication will create numerous mutations and/or chromosome rearrangements—ultimately triggering cell death [3]. Along these lines, DNA topoisomerase inhibitors are of great interest because they help to maintain strand breaks generated by topoisomerases during replication. In this article, we discuss the characteristics of topoisomerase (DNA) I (TOP1) and its inhibitors, as well as the underlying DNA repair pathways and the use of TOP1 inhibitors in cancer therapy.
Collapse
Affiliation(s)
- Yang Xu
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Mail Drop 64-7520, Pullman, WA 99164, USA.
| | - Chengtao Her
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Mail Drop 64-7520, Pullman, WA 99164, USA.
| |
Collapse
|
128
|
Thompson JM, Nguyen QH, Singh M, Razorenova OV. Approaches to identifying synthetic lethal interactions in cancer. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2015; 88:145-55. [PMID: 26029013 PMCID: PMC4445436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Targeting synthetic lethal interactions is a promising new therapeutic approach to exploit specific changes that occur within cancer cells. Multiple approaches to investigate these interactions have been developed and successfully implemented, including chemical, siRNA, shRNA, and CRISPR library screens. Genome-wide computational approaches, such as DAISY, also have been successful in predicting synthetic lethal interactions from both cancer cell lines and patient samples. Each approach has its advantages and disadvantages that need to be considered depending on the cancer type and its molecular alterations. This review discusses these approaches and examines case studies that highlight their use.
Collapse
Affiliation(s)
| | | | | | - Olga V. Razorenova
- Olga V. Razorenova, University of California Irvine, 845 Health Sciences Road, Gross Hall Room 3010, Mail Code 3900, Irvine, CA 92697; Tele: 949-824-8156;
| |
Collapse
|